Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cancer Gene Ther ; 20(12): 651, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24346318
3.
Cancer Gene Ther ; 13(8): 725-31, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16543919

ABSTRACT

This review summarizes clinical experience with infectious disease vaccines and data from animal tumor models that support a paradigm shift for cancer vaccines from therapeutic to prevention applications.


Subject(s)
Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Neoplasms/prevention & control , Algorithms , Animals , Animals, Genetically Modified , Antigens, Neoplasm/immunology , Humans , Neoplasms/therapy , Neoplasms, Experimental/prevention & control , Neoplasms, Experimental/therapy
4.
Clin Exp Immunol ; 129(1): 99-106, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12100028

ABSTRACT

We report the immunological characterization of three colon carcinoma cell lines, COLO 205, SW620 and SW403, which we selected to combine with cytokine-secreting fibroblasts for the development of an allogeneic tumour cell vaccine. The cell lines expressed HLA-A2 as well as shared tumour-associated antigens (TAAs) representative of colon carcinomas: CEA, Ep-CAM, MUC1, HER2/neu and MAGE antigens. They did not secrete high levels of the immunosuppressive factors TGF-beta, IL-10 or prostaglandins. The lines presented TAAs in a manner recognized by immune effector cells, which was demonstrated by the lysis of SW620 by HLA-A2-restricted anti-p53 cytotoxic T lymphocytes (CTL). COLO 205 and SW620 were genetically modified to express the co-stimulatory molecule CD80 (B7.1), which increased the ability of the cells to stimulate CTL in vitro. CTL clones derived from HLA-A2+ peripheral blood mononuclear cells stimulated with the CD80-expressing lines lysed the stimulator cell and an HLA-A2+ colon cancer cell line, but did not lyse an isogeneic fibroblast line or an HLA-A2- colon cancer cell line. CTL clones derived from colon carcinoma patients immunized with an allogeneic vaccine containing these lines demonstrated killing of autologous tumour cells, the vaccine cell lines and other HLA-A2+ colon cancer cell lines, but not fibroblasts isogeneic to certain of the target cell lines. Our studies demonstrate that these colon carcinoma cell lines express shared TAAs that can induce CTLs which recognize and lyse other colon carcinoma cells, and support the continued clinical evaluation of the CD80 gene modified allogeneic colon cell/cytokine-secreting fibroblast carcinoma vaccine.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Colonic Neoplasms/immunology , HLA-A2 Antigen/immunology , Isoantigens/immunology , Tumor Cells, Cultured/immunology , Antigen Presentation , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Carcinoembryonic Antigen/immunology , Cell Adhesion Molecules/immunology , Cells, Cultured , Colonic Neoplasms/prevention & control , Cytokines/metabolism , Cytotoxicity, Immunologic , Epithelial Cell Adhesion Molecule , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Lymphocyte Activation , Mucin-1/immunology , Neoplasm Proteins/immunology , Receptor, ErbB-2/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Transforming Growth Factor beta/metabolism
6.
J Clin Oncol ; 17(10): 3313-23, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10506635

ABSTRACT

PURPOSE: We have completed a phase I study, followed by three phase I/II studies, in patients with metastatic melanoma, renal cell carcinoma (RCC), and sarcoma in order to evaluate the safety, toxicity, and antitumor activity of Leuvectin (Vical Inc, San Diego, CA), a gene transfer product containing a plasmid encoding human interleukin (IL)-2 formulated with the cationic lipid 1, 2-dimyristyloxypropyl-3-dimethyl-hydroxyethyl ammonium bromide/dioleyl-phosphatidyl-ethanolamine (DMRIE/DOPE) and administered intratumorally. PATIENTS AND METHODS: Twenty-four patients were treated in the phase I study. Leuvectin doses were 10 microg, 30 microg, or 300 microg weekly for 6 weeks. In three subsequent phase I/II studies, a total of 52 patients (18 with melanoma, 17 with RCC, and 17 with sarcoma) were treated with further escalating doses of Leuvectin: 300 microg twice a week for 3 weeks, 750 microg weekly for 6 weeks, and 1,500 microg weekly for 6 weeks. RESULTS: There were no drug-related grade 4 toxicities and only one grade 3 toxicity, but the majority of patients experienced mild constitutional symptoms after treatment. In the phase I/II studies, 45 patients were assessable for response (14 with RCC, 16 with melanoma, and 15 with sarcoma). Two patients with RCC and one with melanoma have achieved partial responses lasting from 16 to 19 months and continuing. In addition, two RCC, three melanoma, and six sarcoma patients had stable disease lasting from 3 to 18 months and continuing. The plasmid was detected by polymerase chain reaction assay in the posttreatment samples of 29 of 46 evaluated patients. Immunohistochemistry studies on serial biopsy specimens showed increased IL-2 expression and CD8(+) infiltration after treatment in the tumor samples of several patients (12 and 16, respectively). CONCLUSION: Direct intratumoral injection of Leuvectin is a safe and possibly effective immunotherapeutic approach in the treatment of certain tumor types.


Subject(s)
Carcinoma, Renal Cell/therapy , Gene Transfer Techniques , Genetic Therapy , Interleukin-2/therapeutic use , Kidney Neoplasms/therapy , Melanoma/therapy , Sarcoma/therapy , Skin Neoplasms/therapy , Adult , Aged , CD8 Antigens/analysis , Carcinoma, Renal Cell/pathology , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Interleukin-2/genetics , Interleukin-2/pharmacokinetics , Kidney Neoplasms/pathology , Lipids/genetics , Lipids/therapeutic use , Male , Melanoma/pathology , Middle Aged , Plasmids/genetics , Polymerase Chain Reaction , Quaternary Ammonium Compounds/therapeutic use , Sarcoma/pathology , Skin Neoplasms/pathology
7.
Clin Cancer Res ; 5(9): 2359-65, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499605

ABSTRACT

The purpose of this study was to determine the safety, toxicity, and antitumor immune response following S.C. immunizations with a mixture of irradiated, autologous tumor cells and autologous fibroblasts that were genetically modified to express the gene for interleukin 2 (IL-2) in patients with colorectal carcinoma. Ten patients were treated with a fixed dose of tumor cells (10(7)) and escalating doses of fibroblasts secreting IL-2 (per 24 h): 100 units (three patients), 200 units (three patients), 400 units (three patients), and 800 units (one patient). Pre- and posttreatment peripheral blood mononuclear cells were evaluated for evidence of antitumor immune responses. Fatigue and/or flu-like symptoms were experienced by seven patients and delayed-type hypersensitivity-like skin reactions were observed at the sites of the second or subsequent vaccinations in five patients. Low frequencies of tumor cytotoxic T-cell precursors (range, 1/190,000-1/1,320,000 peripheral blood mononuclear cells) were detected prior to therapy in four of seven patients. There was a 5-fold increase following treatment in the frequency of tumor cytotoxic T-cell precursors in two of six evaluable patients. Some patients with colorectal cancer have low frequencies of tumor cytotoxic T-cell precursors that may be increased by this well-tolerated form of IL-2 gene therapy, which warrants continued clinical evaluation.


Subject(s)
Cancer Vaccines/therapeutic use , Colorectal Neoplasms/therapy , Fibroblasts/metabolism , Genetic Therapy/methods , Immunotherapy, Adoptive/methods , Interleukin-2/biosynthesis , Interleukin-2/genetics , Cancer Vaccines/immunology , Cell Transplantation , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Combined Modality Therapy , Fibroblasts/physiology , Fibroblasts/transplantation , Genetic Engineering , Genetic Therapy/adverse effects , Humans , Hypersensitivity, Delayed/etiology , Hypersensitivity, Delayed/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/radiation effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/radiation effects , T-Lymphocytes, Cytotoxic/transplantation
8.
Gynecol Oncol ; 71(2): 204-10, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9826461

ABSTRACT

The immunosuppressive protein transforming growth factor beta (TGF-beta) inhibits the activation of various immune effector cells including cytotoxic T lymphocytes and may therefore inhibit the efficacy of immunostimulatory interleukin-2 (IL-2) gene therapy. In this study, we investigated the effect of TGF-beta downregulation on IL-2 gene therapy in the intraperitoneal model of murine ovarian teratoma (MOT). MOT cells, like many human ovarian carcinomas, were found to produce TGF-beta. Production of TGF-beta by MOT cells was suppressed using a TGF-beta antisense plasmid vector (pCEP4/TGF-beta antisense). Subcutaneous immunization of C3H mice with a mixture of IL-2 gene-transduced fibroblasts and TGF-beta antisense-modified MOT cells induced significantly better protection against a subsequent intraperitoneal tumor challenge compared with immunization with unmodified MOT cells alone [11/16 (69%) vs 4/21 (19%) tumor-free animals, P < 0.01]. Immunization with either a mixture of IL-2 gene modified fibroblasts and unmodified MOT cells [2/12 (17%) tumor-free animals] or TGF-beta antisense-modified MOT cells alone (0/13 tumor free animals) failed to induce significant protection compared with immunization with unmodified MOT cells. These data show that combined TGF-beta antisense and IL-2 gene therapy is required to generate effective antitumor responses in the MOT model. Our findings suggest that tumor cell expression of immunosuppressive factors may inhibit cytokine immunogene therapy and may have potential implications for the development of future clinical immunogene therapy protocols.


Subject(s)
Antisense Elements (Genetics)/therapeutic use , Cancer Vaccines/therapeutic use , Genetic Therapy , Interleukin-2/genetics , Ovarian Neoplasms/therapy , Teratoma/therapy , Transforming Growth Factor beta/antagonists & inhibitors , 3T3 Cells , Animals , Down-Regulation , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C3H
9.
J Immunother ; 20(6): 437-48, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9409449

ABSTRACT

Several investigators have employed interleukin-2 (IL-2) gene transfer to enhance the immunogenicity of tumor cell vaccines. We describe in this report the construction and characterization of retroviral vectors for IL-2 gene therapy. Human IL-2 cDNA with a chimeric rat preproinsulin/IL-2 DNA leader sequence was subcloned into the pLXSN (long terminal repeat promoter) and pLNCX (cytomegalovirus [CMV] promoter) vectors to generate the plasmids pLXSN-iIL2 and pLNCX-iIL2, respectively. Human IL-2 cDNA with a chimeric human tissue factor/IL-2 DNA leader sequence was utilized to construct the vector pLXSN-tIL2. The levels of IL-2 secreted by transduced tumor cells and fibroblasts were evaluated by enzyme-linked immunosorbent assay (ELISA) of culture supernatants and compared with those of normal peripheral blood mononuclear cells (PBMC) activated in vitro with calcium ionophore and phorbol 12-myristate 13-acetate. The highest levels of IL-2 secreted by transduced tumor cells (760 units/10(6) cells/24 h), adult fibroblasts (625 units/10(6) cells/24 h), and embryonic fibroblasts (3,975 units/10(6) cells/24 h) were 150- to 1,000-fold higher than than secreted by the activated PBMC (4 units/10(6) cells/24 h). Similar levels of IL-2 were expressed by human fibroblasts transduced with pLXSN vectors employing the preproinsulin (pLXSN-iIL2) or tissue factor (pLXSN-tIL2) leader sequences (range in IL-2 units/10(6) cells/24 h pLXSN-iIL2 = 375-625 vs. pLXSN-tIL2 = 90-440). Because IL-2-transduced cells for clinical applications are generally irradiated to prevent cellular proliferation, we evaluated the effects of radiation on IL-2 production. Radiation doses between 1,500 and 10,000 cGy resulted in gradual decreases in IL-2 secretion by transduced cells. The range of the decrease in IL-2 secretion was 7-11% by day 7, 0-29% by day 14, and 25-50% by day 35. For clinical applications, stable production of the vector in high concentrations is an important consideration. The retroviral vector pLXSN-tIL2 produced the highest viral titer and was chosen for further characterization. Southern blot analysis of SacI-digested genomic DNA from the LXSN-tIL2 producer cell line and SacI-digested pLXSN-tIL2 plasmid DNA revealed the expected 3.2-kbp fragment, suggesting the absence of transgene rearrangement and the suitability of this vector as a candidate for clinical applications.


Subject(s)
Genetic Therapy , Genetic Vectors , Interleukin-2/genetics , Retroviridae/genetics , 3T3 Cells , Animals , Base Sequence , Cell Line , Gene Expression , Gene Transfer Techniques , Humans , Insulin , Lymphocytes/metabolism , Mice , Plasmids , Polymerase Chain Reaction , Proinsulin/genetics , Protein Precursors/genetics , RNA, Messenger/analysis , Rats , Thromboplastin/genetics
11.
Proc Natl Acad Sci U S A ; 93(7): 2909-14, 1996 Apr 02.
Article in English | MEDLINE | ID: mdl-8610141

ABSTRACT

Like human gliomas, the rat 9L gliosarcoma secretes the immunosuppressive transforming growth factor beta (TGF-beta). Using the 9L model, we tested our hypothesis that genetic modification of glioma cells to block TGF-beta expression may enhance their immunogenicity and make them more suitable for active tumor immunotherapy. Subcutaneous immunizations of tumor-bearing animals with 9L cells genetically modified to inhibit TGF-beta expression with an antisense plasmid vector resulted in a significantly higher number of animals surviving for 12 weeks (11/11, 100%) compared to immunizations with control vector-modified 9L cells (2/15, 13%) or 9L cells transduced with an interleukin 2 retroviral vector (3/10, 30%) (P < 0.001 for both comparisons). Histologic evaluation of implantation sites 12 weeks after treatment revealed no evidence of residual tumor. In vitro tumor cytotoxicity assays with lymph node effector cells revealed a 3- to 4-fold increase in lytic activity for the animals immunized with TGF-beta antisense-modified tumor cells compared to immunizations with control vector or interleukin 2 gene-modified tumor cells. These results indicate that inhibition of TGF-beta expression significantly enhances tumor-cell immunogenicity and supports future clinical evaluation of TGF-beta antisense gene therapy for TGF-beta-expressing tumors.


Subject(s)
Brain Neoplasms/therapy , DNA, Antisense/therapeutic use , Genetic Therapy , Gliosarcoma/therapy , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Animals , Brain Neoplasms/pathology , Cytotoxicity, Immunologic , DNA, Antisense/administration & dosage , Gene Expression/drug effects , Genetic Therapy/methods , Genetic Vectors , Gliosarcoma/pathology , Humans , Immunotherapy/methods , Interleukin-2/biosynthesis , Lymphocytes/immunology , Plasmids , Rats , Rats, Inbred F344 , Retroviridae , Time Factors
13.
Mol Carcinog ; 14(4): 275-85, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8519417

ABSTRACT

It is known that transfer of the wild-type p53 gene into p53-negative cells from transgenic mice increases their sensitivity to drug and radiation-induced apoptosis. However, unlike many human tumors, these transgenic cells do not express mutant p53, and it is not known from these earlier studies whether wild-type p53 dominates the effects of mutant p53 with respect to drug and radiation sensitivity. We addressed this question in glioblastoma, a disease characterized by an unusually high level of intrinsic resistance to therapy and poor prognosis: mean survival time from diagnosis is only about 1 yr. We introduced the gene for wild-type p53 into human T98G glioblastoma cells, which express endogenous mutant p53 but not wild-type p53. Stable transfectants that co-expressed mutant and wild-type p53 had enhanced sensitivity to cisplatin and gamma radiation, compared with parental cells, control vector-transduced cells, and transduced cells that had lost expression of wild-type p53. Transient wild-type p53 expression after high-efficiency gene transfer by a p53 adenovirus also sensitized the cells to cisplatin and correlated with the induction of apoptosis. The sensitization effect was also observed in p53 adenovirus-infected H23 small cell lung carcinoma cells, which express endogenous mutant p53. Therefore, wild-type p53 gene transfer has dominant effects over mutant p53 in sensitizing tumor cells to therapy, which supports the potential of p53 gene therapy to enhance the efficacy of traditional therapy.


Subject(s)
Gene Transfer Techniques , Genes, p53 , Glioblastoma/genetics , Glioblastoma/therapy , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Base Sequence , Cisplatin/pharmacology , Clone Cells , Drug Screening Assays, Antitumor , Genes, p53/genetics , Glioblastoma/pathology , Humans , Molecular Sequence Data , Mutation , Radiation Tolerance , Transduction, Genetic , Tumor Cells, Cultured
14.
J Immunother Emphasis Tumor Immunol ; 17(4): 201-8, 1995 May.
Article in English | MEDLINE | ID: mdl-7582256

ABSTRACT

We compared the efficacy of gene therapy mediated by interleukin-2 (IL-2) gene-modified tumor cells to gene therapy mediated by IL-2 transduced fibroblasts in the CT-26 model of murine colorectal carcinoma. We transduced CT-26 tumor cells and BALB/c 3T3 fibroblasts with three different retroviral vectors using three different promoters for the human IL-2 gene: DC/TKIL-2 (thymidine kinase promoter), LXSN-iIL2 (long terminal repeat promoter), and LNCX-iIL2 (cytomegalovirus promoter). These transductions resulted in CT-26 and 3T3 subclones that secreted different amounts of IL-2. Immunization of animals with either CT-26/IL-2 cells or with fibroblast/IL-2 cells mixed with CT-26 induced similar levels of immunity that protected 62-82% of animals against a subsequent tumor challenge with parental CT-26. However, mice developed tumors at the site of inoculation in 46% of the animals immunized with CT-26/IL-2 cells. In a separate experiment, CT-26/IL-2 cells were exposed to 6,000 cGy of gamma irradiation to prevent tumor growth at the site of inoculation. Although the CT-26/IL-2 cells continued to secrete IL-2 after irradiation, they were no longer effective at inducing antitumor immunity. In contrast, both irradiated and nonirradiated fibroblast/IL-2 cells, mixed with irradiated CT-26, were equally effective at inducing antitumor immunity. These data suggest that in the CT-26 model, fibroblast-mediated IL-2 gene therapy has advantages for the induction of antitumor immunity and abrogation of tumorigenic potential at the site of inoculation compared with tumor cell-mediated IL-2 gene therapy.


Subject(s)
3T3 Cells/drug effects , Colorectal Neoplasms/therapy , Genetic Therapy , Interleukin-2/genetics , Transfection , 3T3 Cells/radiation effects , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Disease Models, Animal , Humans , Interleukin-2/pharmacology , Interleukin-2/radiation effects , Mice , Mice, Inbred BALB C , Tumor Cells, Cultured
15.
Hum Gene Ther ; 6(5): 591-601, 1995 May.
Article in English | MEDLINE | ID: mdl-7578396

ABSTRACT

We evaluated the effects of different doses of interleukin-2 (IL-2)-transduced fibroblasts in the treatment of colorectal carcinoma in the CT-26 murine tumor model. Immunization with a mixture of irradiated tumor cells and IL-2-transduced fibroblasts (100 units of IL-2/24 hr) induced significantly greater protection against a live tumor challenge compared to irradiated tumor cells alone (22/35, 65% vs. 10/30, 33%, p < 0.02). Protective effects were observed with doses of IL-2-transduced fibroblasts secreting from 5 to 100 units of IL-2/24 hr. Parallel experiments in nude mice produced no protection, indicating that the effects of immunization were mediated by a T-cell-dependent mechanism. In animals with established tumors, complete tumor remissions were observed following immunization with a mixture of irradiated tumor cells and IL-2-transduced fibroblasts secreting 100 units of IL-2/24 hr, but not after immunization with irradiated tumor cells alone (7/16 vs. 0/11 complete remissions, p < 0.02). Fibroblasts secreting higher doses of IL-2 were ineffective in generating systemic immunity, but were required to prevent tumor implantation. A statistically significant difference in the prevention of tumor implantation was observed between groups inoculated with a mixture of live tumor cells and IL-2-transduced fibroblasts (1,750 units of IL-2/24 hr) compared to control fibroblasts (6/8 vs. 0/12, p < 0.001). Similar results were observed in nude mice, suggesting that the implantation rejection response is mediated in part by cells other than thymus-derived T cells. Our data support the utility of IL-2-transduced fibroblasts and indicate that the level of IL-2 expression is an important variable in activating different effector components of antitumor immune responses in IL-2 gene therapy.


Subject(s)
Colorectal Neoplasms/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Interleukin-2/genetics , T-Lymphocytes/immunology , Adenocarcinoma/pathology , Animals , Cell Line , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Cytotoxicity, Immunologic , Fibroblasts , Gene Expression , Genetic Vectors , Humans , Immunity, Cellular , Interleukin-2/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Retroviridae/genetics
16.
Blood ; 85(8): 2025-37, 1995 Apr 15.
Article in English | MEDLINE | ID: mdl-7718875

ABSTRACT

The goal of this phase II multicenter clinical trial was to evaluate a new intensive chemotherapy program for adults with untreated acute lymphoblastic leukemia (ALL) and to examine prospectively the impact of clinical and biologic characteristics on the outcome. One hundred ninety-seven eligible and evaluable patients (16 to 80 years of age; median, 32 years of age) received cyclophosphamide, daunorubicin, vincristine, prednisone, and L-asparaginase; 167 patients (85%) achieved a complete remission (CR), 13 (7%) had refractory disease, and 17 (9%) died during induction. A higher CR rate was observed in younger patients (94% for those < 30 years old, 85% for those 30 to 59 years old, and 39% for those > or = 60 years old, P < .001) and in those who had a mediastinal mass (100%) or blasts with a T-cell immunophenotype. Eighty percent of B-lineage and 97% of T-cell ALL patients achieved a CR (P = .01). The coexpression of myeloid antigens did not affect the response rate or duration. Seventy percent of those with cytogenetic or molecular evidence of the Philadelphia (Ph) chromosome and 84% of those without such evidence achieved a CR (P = .11). Patients in remission received multiagent consolidation treatment, central nervous system prophylaxis, late intensification, and maintenance chemotherapy for a total of 24 months. After a median follow-up time of 43 months, the median survival for all 197 patients is 36 months; the median remission duration for the 167 CR patients is 29 months. Favorable pretreatment characteristics relative to remission duration or survival are younger age, the presence of a mediastinal mass or lymphadenopathy, a white blood cell count (WBC) less than 30,000/microL, L1 morphology, T or TMy immunophenotype, and the absence of the Ph chromosome. The estimates of the proportion surviving at 3 years are 69% for patients less than 30 years old, 39% for those 30 to 59 years old, 89% for those who had a mediastinal mass, 59% with WBC less than 30,000/microL, 63% with L1 morphology, 69% for T or TMy antigen expression, and 62% for those who lack the Ph chromosome. Fifteen patients (8%) had no unfavorable prognostic factors and have an estimated probability of survival at 5 years of 100% (95% confidence interval, 77% to 100%). This intensive chemotherapy regimen produces a high remission rate and a high proportion of durable remissions in adults with ALL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Asparaginase/administration & dosage , Combined Modality Therapy , Cranial Irradiation , Cyclophosphamide/administration & dosage , Cytarabine/administration & dosage , Daunorubicin/administration & dosage , Doxorubicin/administration & dosage , Drug Administration Schedule , Female , Humans , Immunophenotyping , Life Tables , Male , Mercaptopurine/administration & dosage , Methotrexate/administration & dosage , Middle Aged , Multivariate Analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/radiotherapy , Prednisone/administration & dosage , Prospective Studies , Remission Induction , Risk Factors , Survival Analysis , Thioguanine/administration & dosage , Treatment Outcome , Vincristine/administration & dosage
17.
Gene Ther ; 2(2): 164-7, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7719933

ABSTRACT

A patient with glioblastoma multiforme (GBM) who had failed conventional therapy was treated with IL-2 gene therapy. The patient received 10 subcutaneous immunizations with autologous tumor cells and fibroblasts genetically modified to secrete IL-2 by retroviral gene transfer. An antitumor immune response mediated in part by CD8+ cytotoxic T cells was demonstrated with the patient's peripheral blood mononuclear cells. A magnetic resonance imaging (MRI) scan performed 4 weeks after the highest treatment dose revealed marked tumor necrosis. These results support the evaluation of this form of IL-2 gene therapy in additional patients with glioblastoma.


Subject(s)
Brain Neoplasms/therapy , Fibroblasts/metabolism , Fibroblasts/transplantation , Genetic Therapy , Glioblastoma/therapy , Interleukin-2/therapeutic use , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/transplantation , Recombinant Fusion Proteins/therapeutic use , Salvage Therapy , Temporal Lobe , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cells, Cultured/transplantation , Combined Modality Therapy , Fatal Outcome , Female , Genetic Vectors , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Injections, Subcutaneous , Interleukin-2/genetics , Interleukin-2/metabolism , Isotretinoin/therapeutic use , Lomustine/administration & dosage , Magnetic Resonance Imaging , Middle Aged , Necrosis , Neoplasm Recurrence, Local , Procarbazine/administration & dosage , Radioimmunotherapy , Radiosurgery , Recombinant Fusion Proteins/metabolism , Retroviridae/genetics , T-Lymphocytes, Cytotoxic/immunology , Tamoxifen/therapeutic use , Transfection , Tumor Cells, Cultured/transplantation , Vincristine/administration & dosage
20.
Leukemia ; 8(11): 1918-22, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7967737

ABSTRACT

Specific structural rearrangements involving chromosome band 11q23 occur in a variety of hematologic malignancies, including an estimated 2-7% of patients with acute lymphoblastic leukemia (ALL). Translocations involving chromosome band 11q23 have been associated with a poor prognosis in patients with ALL. Recently, a gene known as MLL has been identified which is involved in acute lymphoid and myeloid leukemias with rearrangements at 11q23. A 0.74-kilobase (kb) cDNA probe from the MLL gene can detect both common and uncommon rearrangements involving MLL on conventional Southern blots. We studied 86 newly diagnosed adults entered on an ALL clinical trial to investigate the incidence of MLL gene rearrangements and to determine clinical, morphologic, immunologic and cytogenetic characteristics of such patients. Two of 86 patients had MLL gene rearrangements detected by Southern blot analysis. One of these 86 patients had an 11q23 translocation by cytogenetic analysis whereas the second patient was unevaluable by standard cytogenetic analysis. Southern blot identification of rearrangements involving MLL, especially in patients with limited material for cytogenetic analysis, can provide critical diagnostic and prognostic information which may be useful in the clinical management of patients with these abnormalities.


Subject(s)
Chromosome Aberrations/genetics , DNA-Binding Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogenes , Transcription Factors , Adolescent , Adult , Aged , Chromosome Disorders , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 4 , DNA, Neoplasm/genetics , Female , Gene Rearrangement , Histone-Lysine N-Methyltransferase , Humans , Male , Middle Aged , Myeloid-Lymphoid Leukemia Protein , Translocation, Genetic , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...